EP2623604A1 - Adenoviral-based biological delivery and expression system for use in the treatment of osteoarthritis - Google Patents

Adenoviral-based biological delivery and expression system for use in the treatment of osteoarthritis Download PDF

Info

Publication number
EP2623604A1
EP2623604A1 EP12000703.4A EP12000703A EP2623604A1 EP 2623604 A1 EP2623604 A1 EP 2623604A1 EP 12000703 A EP12000703 A EP 12000703A EP 2623604 A1 EP2623604 A1 EP 2623604A1
Authority
EP
European Patent Office
Prior art keywords
promoter
adenoviral
helper
nucleic acid
dependent
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
EP12000703.4A
Other languages
German (de)
French (fr)
Other versions
EP2623604B1 (en
EP2623604B8 (en
Inventor
Brendan Lee
Kilian Guse
Zhechao Ruan
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Baylor College of Medicine
Original Assignee
Baylor College of Medicine
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to ES12000703.4T priority Critical patent/ES2534782T3/en
Application filed by Baylor College of Medicine filed Critical Baylor College of Medicine
Priority to EP20120000703 priority patent/EP2623604B8/en
Priority to DK12000703.4T priority patent/DK2623604T3/en
Priority to PL12000703T priority patent/PL2623604T3/en
Priority to CN201380007814.1A priority patent/CN104245941B/en
Priority to EA201491457A priority patent/EA033004B1/en
Priority to IN1682KON2014 priority patent/IN2014KN01682A/en
Priority to JP2014555328A priority patent/JP6340320B2/en
Priority to US14/375,351 priority patent/US10301647B2/en
Priority to CA2861408A priority patent/CA2861408C/en
Priority to PCT/IB2013/000198 priority patent/WO2013114199A1/en
Priority to AU2013213873A priority patent/AU2013213873B2/en
Publication of EP2623604A1 publication Critical patent/EP2623604A1/en
Publication of EP2623604B1 publication Critical patent/EP2623604B1/en
Application granted granted Critical
Publication of EP2623604B8 publication Critical patent/EP2623604B8/en
Priority to JP2018016342A priority patent/JP2018075035A/en
Priority to US16/398,691 priority patent/US20190376080A1/en
Priority to US17/497,045 priority patent/US20220073948A1/en
Priority to US17/855,322 priority patent/US20220348963A1/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/545IL-1
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10041Use of virus, viral particle or viral elements as a vector
    • C12N2710/10043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10071Demonstrated in vivo effect
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10341Use of virus, viral particle or viral elements as a vector
    • C12N2710/10343Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10371Demonstrated in vivo effect
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2800/00Nucleic acids vectors
    • C12N2800/24Vectors characterised by the absence of particular element, e.g. selectable marker, viral origin of replication

Definitions

  • the invention relates to the field of genetic engineering and provides an adenoviral-based biological delivery and expression system for use in the treatment of osteoarthritis in human or mammalian joints by long-term gene expression of human or mammalian interleukin-1 receptor antagonist (II-1Ra) in synovial cells.
  • II-1Ra interleukin-1 receptor antagonist
  • Osteoarthritis is a degenerative joint disease that occurs in human or mammalian joints and constitutes a severe economical and medical problem ( Matthews, G.L., and Hunter, D.J. (2011). Emerging drugs for osteoarthritis. Expert Opin. Emerging Drugs 1-13 ., Brooks PM. Impact of osteoarthritis on individuals and society: how much disability? Social consequences and health economic implications. Curr Opin Rheumatol 2002; 14: 573-577 ).
  • Cartilage is the tough connective tissue that covers the ends of bones in joints. It provides for a relatively frictionless, highly lubricated surface between rigid bones and allows for a smooth movement.
  • OA constitutes a significant problem with a tremendous economic impact. Horses spend almost their entire life on their legs, and those used for athletic purposes additionally undergo excessive training. Consequently, most of the joints in athletic horses are heavily overused which often results in lameness. Lameness accounts for about 70 % of the cases where horses cannot participate in races or show jumping. About 60 % of the cases can be directly linked to OA ( Caron JP, Genovese, RL Principals and practices of joint disease treatment. In: Ross MW And Dyson S, eds. Diagnosis and Management of Lameness in the Horse. 1st ed. Philadelphia: Saunders, 2003:746-764 ). Therefore, OA is the most common reason for the inability of a horse to participate in racing competitions or shows.
  • Analgesic treatment usually involves steroids and non-steroidal anti-inflammatory drugs (NSAIDS), which have shown efficacy in the treatment of OA for some decades.
  • NSAIDS non-steroidal anti-inflammatory drugs
  • these drugs can suppress joint inflammation, many of them are known to have deteriorating effects on the cartilage, which further worsens the underlying process of OA development.
  • Hyaluronic acid for instance, which restores viscoelasticity and lubrication of the joints, has also been widely used.
  • polysulphated glycosaminoglycans injected into the joint or intramuscularly as well as orally administered glucosamine and chondroitin sulphate have shown some efficacy, however their mechanisms of action are not fully understood.
  • therapies have only limited efficacy in the treatment of OA and their success often depends on the severity of the case.
  • these drugs must be administered frequently, sometimes even in combination with each other.
  • frequent drug injections into the joint are laborious, bear the risk for infections, cause stress for the horse and are costly.
  • surgery has generally shown low efficacy in horses and is typically only performed in severe advanced-stage subjects. It follows that there is a clear and yet unmet medical need for more efficacious and sustained treatments that are at the same time also cost effective in the long run.
  • interleukin-1 functions as a central mediator of inflammation ( Dinarello CA. Interleukin-1 family. In: Thomson AW, Lotz MT (eds). The Cytokine Handbook. Academic Press; London, 2003, pp 643-668 .;). Moreover, II-1 strongly inhibits matrix synthesis by cartilage and at high concentrations, triggers matrix breakdown ( Evans, C.H., Gouze, J.N., Gouze, E., Robbins, P.D., and Ghivizzani, S.C. (2004). Osteoarthritis gene therapy. Gene Ther 11, 379-389 ).
  • interleukin-1 receptor antagonist II-1Ra
  • II-1Ra interleukin-1 receptor antagonist
  • II-1Ra On nucleic acid level, II-1Ra is considerably conserved among mammalian species.
  • the cDNA sequences of human II-1Ra shares 82% homology with the murine variant (Accession no: NM_031167), 84% with the equine variant (Accession no: NM_001082525). 84% with the canine variant (Accession no, NM_001003096), 84% with the lapine variant (Accession no: NM_001082770) and 82% with the bovine variant (Accession no: NM_174357).
  • the adenoviral vector used for expression of equine II-1Ra DNA was a first-generation adenoviral vector, which was shown to produce biologically active equine II-1Ra.
  • Helper-dependent adenoviruses also known as gutless or high-capacity adenoviruses
  • HDAd gutless or high-capacity adenoviruses
  • adenoviral vectors Mitani, K., Graham, F.L., Caskey, C.T. & Kochanek. S. Rescue, propagation, and partial purification of a helper virus-dependent adenovirus vector.
  • a Helper-dependent adenovirus vector system removal of helper virus by Cre-mediated excision of the viral packaging signal.
  • the patent US 5,747,072 A describes and claims a recombinant adenoviral vector having an expression control sequence operatively linked to a gene that encodes an anti-inflammatory polypeptide, ribozyme or antisense RNA molecule.
  • Administering to the joint a therapeutically effect amount of a recombinant first generation adenoviral vector resulted In a reduced inflammatory response in the joint of the treated subject. Again, as in other studies, the long-term expression of II-1Ra was limited.
  • II-1Ra biologically active recombinant interleukin-1 receptor antagonist
  • the solution for the problem is provided by an adenoviral-baaed biological delivery and expression system having the features as claimed in claim 1.
  • Preferred embodiments of the invention are subject-matter of the dependent claims.
  • the adenoviral-based biological delivery and expression system is based on a helper-dependent adenoviral vector containing a nucleic acid sequence encoding for human or mammalian interleukln-1 receptor antagonist (II-1Ra), left and right inverted teminal repeats (L ITR and R ITR), adenoviral packaging signal and non-viral, non-coding stuffer nucleic acid sequences.
  • II-1Ra interleukln-1 receptor antagonist
  • L ITR and R ITR left and right inverted teminal repeats
  • adenoviral packaging signal and non-viral, non-coding stuffer nucleic acid sequences.
  • the gene of the human or mammalian interieukin-1 receptor antagonist (II-1Ra) is controlled by an inflammation-inducible promoter
  • Preferred inflammation-regulated promoters for use in the present invention are NF- ⁇ B promoter, interleukin 6 (II-6) promoter, interleukin-1 (II-1) promoter, tumor necrosis factor (TNF) promoter, cyclooxygenase 2 (COX-2) promoter, complement factor 3 (C3) promoter, serum amyloid A3 (SAA3) promoter, macrophage inflammatory protein-1 ⁇ (MIP-1 ⁇ ) promoter, or hybrid constructs of the above.
  • the promoter sequences are upstream of the reading frame of the cloned II-1Ra gene.
  • the inflammation-regulated promoter according to the present invention is specifically activated by increased levels of immune stimulatory substances such as lipopolysaccharide (LPS), which is a major component of the outer cell membrane of grarn-negative bacteria.
  • immune stimulatory substances such as lipopolysaccharide (LPS)
  • LPS lipopolysaccharide
  • a vartiety of immune stimulatory substances and cytokines are released, resulting in high levels of promoter-activating substances.
  • One component, for instance, is NF- ⁇ B, which regulates the NF- ⁇ B promoter. Therefore, the release of such usteoarthritic-specific activators allows for the control of gene expression in joints of humans or mammals for treating or preventing an osteoarthritic condition.
  • an inflammation-inducibie promoter provides for specific control of II-1Ra gene expression in osteoarthritic tissue cells. Only cells that are affected by the disease will express and secrete the II-1Ra gene product, whereas cells that are not affected remain silent.
  • the use of NF- ⁇ B5-ELAM promoter for inflammation-dependent gene expression is most preferred. However, any other inflammafion-dependent promoter that result in a specific expression of the II-1Ra gene product in osteoarthritic tissue can be used in context of the present invention.
  • helper-dependent adenoviral vector as used In the present invention minimizes immune responses in the host, and confers long-term gene expression of human or mammalian II-1Ra in joints that are affected by osteoarthritis.
  • the helper-dependent adenoviral vector of the invention is preferably administered in a single injection dose directly into the joints of the osteoarthritic subjects.
  • the gene of II-1Ra is delivered to synovial cells such as synoviocytes.
  • synovial cells such as synoviocytes.
  • Synovial cells that are affected by inflammation start to produce recombinant II-1Ra protein under the control of the inflammation-inducible promoter such as the NF- ⁇ B promoter.
  • High amounts of II-1Ra are then secreted into the joint space, where II-1Ra is able to inhibit inflammation and stop cartilage degradation by blocking the interleukln-1 receptor on the surface of synoviocytes and the cells embedded in the cartilage.
  • high local concentrations of recombinant II-1Ra do not show any side effects.
  • adenoviral-based biological delivery and expression system according to the present invention.
  • High local and low systemic concentrations of the therapeutic protein II-1Ra are achieved, resulting in maximum efficacy in the treatment of OA at no or minimal side effects.
  • cells containing the helper-dependent adenoviral vector of the invention are capable to produce recombinant II-1Ra for an extended period of at least one year. Consequently, medical and economic burden associated with frequent joint injections that were required in the known short-term treatments will be significantly reduced.
  • the serious long-term side effects of commonly used steroids can be avoided by using the helper-dependent adenoviral vector of the invention due to its cartilage protective effect.
  • common complications associated with OA treatment are minimized and joint health will be preserved in the long run resulting in sustained health improvement of the treated animal or human.
  • the inflammation-dependent II-1Ra production of the vector of the invention allows for the prevention of the development of an osteoarthritic condition as synovial cells that are infected with the adenoviral vector of the invention remain silent in the absence of immure stimulatory substances that could activate the NF- ⁇ B5-ELAM promoter or any other inflammation-dependent promoter. Only if the osteoarthritic condition initiates, the promoter is activated as a result of inflammation and subsequently II-1Ra is produced and secreted.
  • this mechanism allows for the prevention of the development of osteoarthritis in an early stage.
  • An inflammation-dependent II-1Ra production of the vector of the invention can also be viewer as a safety feature to ensure that II-1Ra is no longer produced, for example when the osteoarthritic condition is resolved or has disappeared.
  • helper-dependent adenoviral vector as used in the present invention does not carry any viral sequences, except the left and right inverted terminal repeats (ITRs) and the adenoviral packaging signal.
  • Preferred helper-dependent adenoviral vectors to be used in the present invention are those based on the helper virus and helper-dependent backbone system developed by Palmer and Ng ( Palmer, D., and Ng, P. (2003). Improved system for helper-dependent adenoviral vector production. Mol Ther 8, 846-852 .) and Toietta et al ( Toietta, G., Pastore, L., Cerullo, V., Finegold, M., Beaudet, A.L., and Lee, B. (2002).
  • a preferred adenoviral delivery and expression system comprises a nucleic acid sequens set forth in SEQ ID NO 2 or SEQ ID NO3, or a biologically effective part thereof.
  • the nucleic acid sequence of SEQ ID NO 2 describes a murine helper-dependent adenoviral vector
  • the sequence set forth in SEQ ID NO 3 describes a equine helper-dependent adenoviral vector, both bearing the murine and equine II-1Ra gene, respectively.
  • the system of the invention has at least 50%, 60%, 70%, 80%, 90% sequence homology with the vector set forth in SEQ ID NO 2 or SEQ ID NO 3.
  • Bioly effective in the context of the present invention means that the gene product of the adenoviral delivery and expression system comprises the full or partial polypeptide sequence of II-1Ra having the in-joint activity to neutralize the effect of II-1 on synovial inflammation.
  • the helper-dependent adenoviral vector of the invention preferably contains the cDNA sequence of II-1Ra that is controlled by the inflammation-inducible promoter.
  • II-1Ra contains species-specific nucleic acid sequences
  • the adenoviral vector is able to express interleukin-1 receptor antagonist (II-1Ra) from any mammalian species or human.
  • the cDNA of the mammalian interleukin-1 receptor antagonist (II-1Ra) used for cloning is a cDNA selected from the group consisting of murine II-1Ra, equine II-1Ra, canine II-1Ra, cat II-1Ra, rabbit II-1Ra, hamster II-1Ra, bovine II-1Ra, camel II-1Ra or their homologs in other mammalian species.
  • helper-dependent adenoviral vector preferably further comprises a marker gene that is visually or instrumentally detectable.
  • Preferred marker genes are, for instance, green fluorescence protein (GFP) or luciferase enzyme.
  • nucleic acid sequence of murine II-1Ra as used in the present invention is shown in the sequence listing set forth in SEQ ID NO 1.
  • any nucleic acid sequence resulting in a biologically active II-1Ra protein of any mammalian or human species can be used in the context of the present invention.
  • conserved nucleic acid sequences encoding for the same amino adds, polypeptide or protein fall under scope of the present invention.
  • II-1Ra genes of various species share a high homology among each other.
  • the helper-dependent adenoviral vector according to the invention contains a nucleic acid sequence (e.g.
  • the invention also comprises biologically active nucleic acid sequences of II1-Ra or fragments thereof beside the full-length nucleic acid sequence of II1Ra.
  • the present invention further comprises a pharmaceutical composition, comprising a helper-dependent adenoviral vector containing a nucleic acid sequence encoding for human or mammalian interleukin-1 receptor antagonist (II-1Ra), left and right inverted terminal repeats (L ITR and R ITR), packaging signal and non-viral, non-coding stuffer nucleic acid sequences, wherein the expression of the human or mammalian interiaukin-1 receptor antagonist (II-1Ra) gene within synovial cells is regulated by an inflammallon-inducible promoter, for the treatment or prevention of osteoathritis.
  • a helper-dependent adenoviral vector containing a nucleic acid sequence encoding for human or mammalian interleukin-1 receptor antagonist (II-1Ra), left and right inverted terminal repeats (L ITR and R ITR), packaging signal and non-viral, non-coding stuffer nucleic acid sequences, wherein the expression of the human or mammalian interiaukin
  • Preferred promoters as used in the context of the present invention are NF- ⁇ B promoter, interleukin 6 (II-6) promoter, interleukin-1 (II-1) promoter, tumor necrosis factor (TNF) promoter, cyclooxygenase 2 (COX-2) promoter, complement factor 3 (C3) promoter, serum amyloid A3 (SAA3) promoter, macrophage inflammatory protein-1 ⁇ (MIP-1 ⁇ ) promoter, or hybrid constructs of the above.
  • II-6 interleukin 6
  • II-1 interleukin-1
  • TNF tumor necrosis factor
  • COX-2 cyclooxygenase 2
  • COX-2 complement factor 3
  • SAA3 serum amyloid A3
  • MIP-1 ⁇ macrophage inflammatory protein-1 ⁇
  • the adenoviral delivery and expression system specifically locates in the joints when administered intra-atrticularly. Most importantly, no measurable concentration of vector sequences could be deteced in the liver of mice treated with the adenoviral system of the invention. Therefore, II-1Ra concentrations are expected to be highest in the joints injected with the vector of the invention while no significant side effects are expected in any other organ.
  • Figure 1 shows gene maps of the HDAd vectors of the invention.
  • the full vector sequence is shown in SEQ ID NO 2 OR SEQ ID NO 3.
  • GQ-201 carries the equine variant of II-1Ra
  • HDAd-mll-1Ra has the murine II-1Ra variant.
  • Both vectors contain the inflammation inducible NF- ⁇ B5-ELAM promoter upstream of the II-1Ra cDNA according to SEQ ID NO 1 as well as inverted terminal repeats (ITR) and an adenoviral packaging signal.
  • ITR inverted terminal repeats
  • the vectors were cloned by standard digestion/ligation reactions according to the following strategy.
  • the luciferase cDNA in pNifty-luc a plasmid that contains the luciferase cDNA driven by a NF- ⁇ B5-ELAM promoter, was excised with Ncol and Nhel and cDNAs for equine or murine II-1Ra were ligated into this position.
  • the NF- ⁇ B5-ELAM promoter - murine II-1Ra or NF- ⁇ B5-ELAM promoter-equine II-1Ra cassettes were excised with NotI and PacI or EcoRI and PacI. blunted and inserted into pLPBL shuttle plasmid, which had been linearized with SalI and blunted.
  • NF- ⁇ B5-ELAM promoter- murine II-1Ra or NF- ⁇ B5-ELAM promoter - equine II-1Ra cassettes were then excised with AscI, which flanks both sides of the multiple cloning site, and ligated into AscI linearized p ⁇ 28 plasmid ( Toietta, G., Pastore, L., Cerullo, V., Finegold, M., Beaudet, A.L., and Lee, B. (2002). Generation of helper-dependent adenoviral vectors by homologous recombination.
  • HDAd mediates long-term marker gene expression in joints
  • mice were infected intra-articularly with a helper-dependent adenoviral vector of the invention (HDAd) and, for comparison, a first generation adenovirus (Ad) vector expressing firefly luciferase (luc) under the control of a CMV promoter.
  • Ad helper-dependent adenoviral vector of the invention
  • luc firefly luciferase
  • Luc expression was followed over time using in vivo bioluminescence imaging. Strong initial luc signals were detected three days after injection with both vectors ( Figure 2A ). Expression decreased with both vectors thereafter and was undetectable after one month with the first generation vector Ad-luc ( Figure 2B ).
  • HDAd-luc luciferase expression stabilized at day 10 and has been at this level for 380 days.
  • mice were injected intra-articularly with a LacZ expressing HDAd. Strong LacZ expression was seen in the synovium, however, no expression could be observed in chondrocytes ( Figure 3 ).
  • the inventors also analyzed the liver of these animals to assess whether virus escapes from the joints or is spilled during the injection. Most importantly, no detectable vector concentrations over background could be measured by quantitative PCR (data not shown). Therefore, the vector specifically locates in tne joints and remains there, which is of great benefit in the treatment or prevention of an osteoarthritic condition since it suggests minimal side effects.
  • HDAd-II-1Ra prevents the development of OA in mice
  • the model involves transection of anterior and posterior cruciate ligaments of the knee joints, which leads to development of severe OA.
  • Mice were sacrificed one month after OA induction and joints were prepared histologically and stained with Safranin O.
  • the development of OA was scored by a blinded pathologist according to OARSI (Osteoarthritis Research Society International) standard (assignment of scores on a scale of 1-6, 1: no signs of OA at all, 6: maximum OA).
  • HoAd-II-1Ra treated joints had significantly lower OA scores than HDAd-GFP treated or untreated joints, suggesting that HDAd-II1Ra prevented the development of OA ( Figure 5 ).
  • the control vector HDAd-GFP did not seem to have any effect on the development of OA since the average OA score was comparable to the score of the untreated group.
  • HDAd-mII-Ra treats OA in a murine model of the disease
  • HDAd-mII-1Ra The efficacy of HDAd-mII-1Ra in the treatment of OA was evaluated in the murine disease model described above.
  • the model was used to assess whether HDAd-II1-Ra can efficiently treat OA. Therefore, OA was induced by cruciate ligament transection (except in the untransected group) and OA was allowed to develop for two weeks.
  • HDAd-II-1Ra, the control vector (HDAd-GFP) or vehicle was then injected and mice were sacrificed to analyze the joints another six weeks later. HDAd-GFP treated and uninjected mice developed OA to the same extent with an average score of approximately 4.5 ( Figure 6A ).
  • HDAd-II-1Ra treated mice had significantly lower OA scores compared with HDAd-GFP and mock treated. No significant difference was found between HDAd-II-1Ra and untransected (OA-free) mice suggesting efficient treatment of the disease or its prevention.
  • the inventors further evaluated the joints in this experiment by micro computer tomography ( ⁇ CT) analysis. This technique combines high resolution (down to 0.5 micron) x-ray CT scanning with phase contrast optics, which enables visualization of cartilage in small animal joints. Three-dimensional reconstruction of joints and computational tissue analysis tools can be used to quantify several cartilage parameters such as volume and surface area. HDAd-II-1Ra treated joints demonstrated significantly higher cartilage volume compared with HDAd-GFP and mock treated joints ( Figure 6B ).
  • the Figure shows a basic gene map of the helper-dependent adenoviral vector of the invention.
  • the vector backbone consists of the left and right inverted terminal repeats (ITR), adenoviral packaging signal ( ⁇ ) and non-coding, non-viral stuffer sequences (remaining unmarked sequence between ITRs).
  • ITR inverted terminal repeat
  • adenoviral packaging signal
  • non-coding, non-viral stuffer sequences residual unmarked sequence between ITRs.
  • the cDNA of murine II-1Ra is cloned between the viral left and right ITRs of the used adenoviral vector.
  • the gene of II1-Ra is controlled by inflammation-inducible NF- ⁇ B5-ELAM promoter.
  • helper-dependent and first generation adenoviral vectors mediate the same level of marker gene expression.
  • Mice were injected intra-articularly with 10 8 virus particles (VP) of a luciferase expressing helper-dependent (HDAd-luc) or a respective first generation (Ad-luc) adenoviral vector.
  • VP virus particles
  • Ad-luc helper-dependent
  • Three days later mice were imaged using IVIS 200 series Imaging system (Caliper Life Sciences, Hopkintom MA). Strong bioluminescence signals were detected in the joints injected with both HDAd-luc and Ad-luc adenoviral vector. Both knee joints of four mice per group were injected; representative pictures of two mice of each group are shown.
  • Helper-dependent adenoviral vector mediates long-term marker gene expression in joints. Luciferase expression of the mice described in A was followed by repeated bioluminescence imaging and quantified using Living Image 2.5 software (Caliper Life Sciences). Expression decreased and was undetectable by 30 days with the first generation adenoviral vector (Ad-lue). With the helper-dependent adenoviral vector (HDAd-luc) expression also declined but plateaued at 10 days and has been around this level for 380 days.
  • Helper-dependent adenoviral vector infects synoviocytes efficiently. Mice were Injected intra-articularly with 10 8 VP of a LacZ expressing HDAd. One day later, mice were sacrificed and LacZ slaining on sectioned joints was performed. Strong expression (dark blue staining) was seen in the synovium while no staining could be observed in chondrocytes. Right picture is a higher magnification photograph (40x) of the framed area in the left picture (5x).
  • LPS lipopolysaccharides
  • HDAd-II-1Ra samples Increased to about 1600 pg/ml whereas uninduced cells produced less II-1Ra compared to the previous day. No II-1Ra expression was detected in any of the control samples (HDAd-GFP and mock).
  • HDAd-II-1Ra prevents the development of OA.
  • Mice were injected intra-articularly into the knee joints with 10 8 VP of HDAd-II-1Ra, HDAd-GFP or mock and OA was induced by cruciate ligament transduction two days later. Mice were sacrificed after 4 weeks and joints were histologically prepared, sectioned and stained with Safranin O.
  • a blinded pathologist evaluated the level of OA according to OARSI (Osteoarthritis Research Society International) standards (assignment of scores on a scale of 1-6, 1: no signs of OA at all, 6: maximum OA).
  • HDAd-II-1Ra efficiently treats OA in mice.
  • a blinded pathologist evaluated the level of OA according to OARSI (Osteoarthritis Research Society International) standard (assignment of scores on a scale of 1-6, 1: no signs of OA at all, 6: maximum OA).
  • B. HDAd-II-1Ra treated joints demonstrate significantly higher cartilage volume compared to controls.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Biomedical Technology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Molecular Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Virology (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Immunology (AREA)
  • Toxicology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • General Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Rheumatology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

The invention relates to an adenoviral-based biological delivery and expression system for use in the treatment or prevention of osteoathritis in human or mammalian joints by long-term inducible gene expression of human or mammalian interleukin-1 receptor antagonist (II-1Ra) in synovial cells, comprising a helper-dependent adenoviral vector containing a nucleic acid sequence encoding for human or mammalian interleukin-1 receptor antagonist (II-1Ra), left and right inverted terminal repeats (L ITR and R ITR), the adenoviral packaging signal and non-viral, non-coding stuffer nucleic acid sequences, wherein the expression of the human or mammalian interleukin-1 receptor antagonist (II-1Ra) gene within synovial cells is regulated by an inflammation-inducible promoter.

Description

  • The invention relates to the field of genetic engineering and provides an adenoviral-based biological delivery and expression system for use in the treatment of osteoarthritis in human or mammalian joints by long-term gene expression of human or mammalian interleukin-1 receptor antagonist (II-1Ra) in synovial cells.
  • Osteoarthritis (OA) is a degenerative joint disease that occurs in human or mammalian joints and constitutes a severe economical and medical problem (Matthews, G.L., and Hunter, D.J. (2011). Emerging drugs for osteoarthritis. Expert Opin. Emerging Drugs 1-13., Brooks PM. Impact of osteoarthritis on individuals and society: how much disability? Social consequences and health economic implications. Curr Opin Rheumatol 2002; 14: 573-577). Cartilage is the tough connective tissue that covers the ends of bones in joints. It provides for a relatively frictionless, highly lubricated surface between rigid bones and allows for a smooth movement. During OA development, cartilage is partially or completely lost due to abnormal or excessive wearing, which leads to exposed bone ends that rub against each other resulting in inflammation, pain, swelling or loss of mobility. By now, the detailed reasons for the initial cartilage loss that leads to OA are not known, but there is a strong correlation between the incidence and age, obesity and joint overuse such as excessive athletic activity. Accordingly, OA is a major problem not only in humans, but in many mammals, in particular in horses that join in racings and show jumping.
  • Especially in horses, OA constitutes a significant problem with a tremendous economic impact. Horses spend almost their entire life on their legs, and those used for athletic purposes additionally undergo excessive training. Consequently, most of the joints in athletic horses are heavily overused which often results in lameness. Lameness accounts for about 70 % of the cases where horses cannot participate in races or show jumping. About 60 % of the cases can be directly linked to OA (Caron JP, Genovese, RL Principals and practices of joint disease treatment. In: Ross MW And Dyson S, eds. Diagnosis and Management of Lameness in the Horse. 1st ed. Philadelphia: Saunders, 2003:746-764). Therefore, OA is the most common reason for the inability of a horse to participate in racing competitions or shows.
  • No curative treatment is currently available for OA - neither for horses, humans nor for any other mammalian species. Medical treatment is mostly aimed at alleviating the symptoms using analgesic drugs rather than establishing worn away cartilage. An analgesic treatment usually involves steroids and non-steroidal anti-inflammatory drugs (NSAIDS), which have shown efficacy in the treatment of OA for some decades. However, while these drugs can suppress joint inflammation, many of them are known to have deteriorating effects on the cartilage, which further worsens the underlying process of OA development. Hyaluronic acid, for instance, which restores viscoelasticity and lubrication of the joints, has also been widely used. Furthermore, polysulphated glycosaminoglycans injected into the joint or intramuscularly as well as orally administered glucosamine and chondroitin sulphate have shown some efficacy, however their mechanisms of action are not fully understood. Thus, currently used therapies have only limited efficacy in the treatment of OA and their success often depends on the severity of the case. Moreover, these drugs must be administered frequently, sometimes even in combination with each other. However, frequent drug injections into the joint are laborious, bear the risk for infections, cause stress for the horse and are costly. In addition, surgery has generally shown low efficacy in horses and is typically only performed in severe advanced-stage subjects. It follows that there is a clear and yet unmet medical need for more efficacious and sustained treatments that are at the same time also cost effective in the long run.
  • During OA, interleukin-1 (II-1) functions as a central mediator of inflammation (Dinarello CA. Interleukin-1 family. In: Thomson AW, Lotz MT (eds). The Cytokine Handbook. Academic Press; London, 2003, pp 643-668.;). Moreover, II-1 strongly inhibits matrix synthesis by cartilage and at high concentrations, triggers matrix breakdown (Evans, C.H., Gouze, J.N., Gouze, E., Robbins, P.D., and Ghivizzani, S.C. (2004). Osteoarthritis gene therapy. Gene Ther 11, 379-389). To neutralize the effect of II-1 on synovial inflammation, treatment with interleukin-1 receptor antagonist (II-1Ra) constitutes a promising concept for treatment of affected osteoarthritic joints (Evans, C.H., Gouze, J.N., Gouze, E., Robbins, P.D., and Ghivizzani, S.C. (2004). Osteoarthritis gene therapy. Gene Ther 11, 379-389.; Caron JP et al. Chondroprotective effect of intraarticular injections of interleukin-1 receptor antagonist in experimental osteoarthritis. Suppression of collagenase-1 expression. Arthritis Rheum 1996; 39: 1535-1544). On nucleic acid level, II-1Ra is considerably conserved among mammalian species. For example, the cDNA sequences of human II-1Ra (Accession no: NM_173842) shares 82% homology with the murine variant (Accession no: NM_031167), 84% with the equine variant (Accession no: NM_001082525). 84% with the canine variant (Accession no, NM_001003096), 84% with the lapine variant (Accession no: NM_001082770) and 82% with the bovine variant (Accession no: NM_174357).
  • The basic concept of using gene therapy for the treatment of arthritis is well established (Evans CH, Robbins PD. Gene therapy for arthritis, In: Wolff JA (ed.). Gene Therapeutics: Methods and Applications of Direct Gene Transform. Birkhauser Boston, 1994. pp 320-343). In the closest prior art, the treatment of equine osteoarthritis by in vivo delivery of the equine interleukin-1 receptor antagonist gene using an adenoviral-mediated gene transfer has been described (D.D. Frisbie, S.C. Ghivizzani, P.D. Robbins, C.H. Evans. C.W. Mcllwraith, Gene Ther 9, 12-20 (2002). The adenoviral vector used for expression of equine II-1Ra DNA was a first-generation adenoviral vector, which was shown to produce biologically active equine II-1Ra. Although clinical examinations of the horses in this study indicated that the therapeutic expression of II-1Ra significantly decreased signs of joint pain as measured by the degree of lameness, the effect of delivery and expression of biologically active equine II-1Ra transgene was only short-term. Already 30 days following treatment of horses with equine II-1Ra by intra-articular injectior, of various amounts of the vector bearing II1-Ra, expression of equine II-1Ra in joints dropped to normal levels. Similar results were also detected in the US 200310091536 A1 , which describes adenovirus particles encoding an interleukin-1 receptor antagonist for use in the treatment of joint disease. The adenovirus particles used were first generation adenoviral vectors.
  • Helper-dependent adenoviruses (HDAd), also known as gutless or high-capacity adenoviruses, are the latest generation of adenoviral vectors (Mitani, K., Graham, F.L., Caskey, C.T. & Kochanek. S. Rescue, propagation, and partial purification of a helper virus-dependent adenovirus vector. Proc Natl Acad Sci USA 92, 3854-3858 (1995): Parks, R.J. et al. A Helper-dependent adenovirus vector system: removal of helper virus by Cre-mediated excision of the viral packaging signal. Proc Natl Acad Sci USA 93, 13565-13570 (1996); Parks, R.J. Improvements In adenoviral vector technology: overcoming barriers for gene therapy. Clin. Genet. 58, 1-11 (2000)). These vectors are devoid of all viral sequences and are able to mediate long-term gene expression in various tissues (e.g. 7 years in the liver) in contrast to the more immunogenic first generation adenoviruses (Brunetti-Pierri, N. et al. Multi-Year Transgene Expression in Nonhuman Primates Following Hepatic Transduction with Helper-Dependent Adenoviral Vectors. American Society of Gene & Cell Therapy, Annual Meeting 2011 Molecular Therapy Volume 19. ). However, longevity of helper-dependent adenoviruses mediated gene expression in joints has not been evaluated to date.
  • The patent US 5,747,072 A describes and claims a recombinant adenoviral vector having an expression control sequence operatively linked to a gene that encodes an anti-inflammatory polypeptide, ribozyme or antisense RNA molecule. Administering to the joint a therapeutically effect amount of a recombinant first generation adenoviral vector resulted In a reduced inflammatory response in the joint of the treated subject. Again, as in other studies, the long-term expression of II-1Ra was limited.
  • It is therefore an object of the present invention to provide an improved delivery and expression system that allows for a long-term expression of biologically active recombinant interleukin-1 receptor antagonist (II-1Ra) in synovial cells at human or mammalian joints for treatment and prevention of osteoarthritis. The solution for the problem is provided by an adenoviral-baaed biological delivery and expression system having the features as claimed in claim 1. Preferred embodiments of the invention are subject-matter of the dependent claims.
  • The adenoviral-based biological delivery and expression system according to the present invention is based on a helper-dependent adenoviral vector containing a nucleic acid sequence encoding for human or mammalian interleukln-1 receptor antagonist (II-1Ra), left and right inverted teminal repeats (L ITR and R ITR), adenoviral packaging signal and non-viral, non-coding stuffer nucleic acid sequences. Furthermore, the gene of the human or mammalian interieukin-1 receptor antagonist (II-1Ra) is controlled by an inflammation-inducible promoter, Preferred inflammation-regulated promoters for use in the present invention are NF-κB promoter, interleukin 6 (II-6) promoter, interleukin-1 (II-1) promoter, tumor necrosis factor (TNF) promoter, cyclooxygenase 2 (COX-2) promoter, complement factor 3 (C3) promoter, serum amyloid A3 (SAA3) promoter, macrophage inflammatory protein-1α (MIP-1α) promoter, or hybrid constructs of the above. The promoter sequences are upstream of the reading frame of the cloned II-1Ra gene. The inflammation-regulated promoter according to the present invention is specifically activated by increased levels of immune stimulatory substances such as lipopolysaccharide (LPS), which is a major component of the outer cell membrane of grarn-negative bacteria. During osteoarthritis, a vartiety of immune stimulatory substances and cytokines are released, resulting in high levels of promoter-activating substances. One component, for instance, is NF-κB, which regulates the NF-κB promoter. Therefore, the release of such usteoarthritic-specific activators allows for the control of gene expression in joints of humans or mammals for treating or preventing an osteoarthritic condition.
  • The use of an inflammation-inducibie promoter provides for specific control of II-1Ra gene expression in osteoarthritic tissue cells. Only cells that are affected by the disease will express and secrete the II-1Ra gene product, whereas cells that are not affected remain silent. The use of NF-κB5-ELAM promoter for inflammation-dependent gene expression is most preferred. However, any other inflammafion-dependent promoter that result in a specific expression of the II-1Ra gene product in osteoarthritic tissue can be used in context of the present invention.
  • The helper-dependent adenoviral vector as used In the present invention minimizes immune responses in the host, and confers long-term gene expression of human or mammalian II-1Ra in joints that are affected by osteoarthritis.
  • To be most effective, the helper-dependent adenoviral vector of the invention is preferably administered in a single injection dose directly into the joints of the osteoarthritic subjects. Following intra-articular injection, the gene of II-1Ra is delivered to synovial cells such as synoviocytes. Synovial cells that are affected by inflammation start to produce recombinant II-1Ra protein under the control of the inflammation-inducible promoter such as the NF-κB promoter. High amounts of II-1Ra are then secreted into the joint space, where II-1Ra is able to inhibit inflammation and stop cartilage degradation by blocking the interleukln-1 receptor on the surface of synoviocytes and the cells embedded in the cartilage. Most importantly, high local concentrations of recombinant II-1Ra do not show any side effects.
  • Therefore, as shown in the examples below, pain, inflammation and cartilage degradation are inhibited effectively using the adenoviral-based biological delivery and expression system according to the present invention. High local and low systemic concentrations of the therapeutic protein II-1Ra are achieved, resulting in maximum efficacy in the treatment of OA at no or minimal side effects. It is further exemplified that cells containing the helper-dependent adenoviral vector of the invention are capable to produce recombinant II-1Ra for an extended period of at least one year. Consequently, medical and economic burden associated with frequent joint injections that were required in the known short-term treatments will be significantly reduced. In particular, the serious long-term side effects of commonly used steroids can be avoided by using the helper-dependent adenoviral vector of the invention due to its cartilage protective effect. Thus, common complications associated with OA treatment are minimized and joint health will be preserved in the long run resulting in sustained health improvement of the treated animal or human.
  • In addition, the inflammation-dependent II-1Ra production of the vector of the invention allows for the prevention of the development of an osteoarthritic condition as synovial cells that are infected with the adenoviral vector of the invention remain silent in the absence of immure stimulatory substances that could activate the NF-κB5-ELAM promoter or any other inflammation-dependent promoter. Only if the osteoarthritic condition initiates, the promoter is activated as a result of inflammation and subsequently II-1Ra is produced and secreted. Thus by using the adenoviral delivery and expression system of the invention, this mechanism allows for the prevention of the development of osteoarthritis in an early stage.
  • An inflammation-dependent II-1Ra production of the vector of the invention can also be viewer as a safety feature to ensure that II-1Ra is no longer produced, for example when the osteoarthritic condition is resolved or has disappeared.
  • The helper-dependent adenoviral vector as used in the present invention does not carry any viral sequences, except the left and right inverted terminal repeats (ITRs) and the adenoviral packaging signal. Preferred helper-dependent adenoviral vectors to be used in the present invention are those based on the helper virus and helper-dependent backbone system developed by Palmer and Ng (Palmer, D., and Ng, P. (2003). Improved system for helper-dependent adenoviral vector production. Mol Ther 8, 846-852.) and Toietta et al (Toietta, G., Pastore, L., Cerullo, V., Finegold, M., Beaudet, A.L., and Lee, B. (2002). Generation of helper-dependent adenoviral vectors by homologous recombination. .). A preferred adenoviral delivery and expression system according to the present invention comprises a nucleic acid sequens set forth in SEQ ID NO 2 or SEQ ID NO3, or a biologically effective part thereof. The nucleic acid sequence of SEQ ID NO 2 describes a murine helper-dependent adenoviral vector, and the sequence set forth in SEQ ID NO 3 describes a equine helper-dependent adenoviral vector, both bearing the murine and equine II-1Ra gene, respectively. Preferably, the system of the invention has at least 50%, 60%, 70%, 80%, 90% sequence homology with the vector set forth in SEQ ID NO 2 or SEQ ID NO 3.
  • "Biologically effective" in the context of the present invention means that the gene product of the adenoviral delivery and expression system comprises the full or partial polypeptide sequence of II-1Ra having the in-joint activity to neutralize the effect of II-1 on synovial inflammation.
  • The helper-dependent adenoviral vector of the invention preferably contains the cDNA sequence of II-1Ra that is controlled by the inflammation-inducible promoter. Although II-1Ra contains species-specific nucleic acid sequences, the adenoviral vector is able to express interleukin-1 receptor antagonist (II-1Ra) from any mammalian species or human. Preferably, the cDNA of the mammalian interleukin-1 receptor antagonist (II-1Ra) used for cloning is a cDNA selected from the group consisting of murine II-1Ra, equine II-1Ra, canine II-1Ra, cat II-1Ra, rabbit II-1Ra, hamster II-1Ra, bovine II-1Ra, camel II-1Ra or their homologs in other mammalian species.
  • In order to monitor the presence of genomic vector sequences in synovial cells, the helper-dependent adenoviral vector according to the invention preferably further comprises a marker gene that is visually or instrumentally detectable. Preferred marker genes are, for instance, green fluorescence protein (GFP) or luciferase enzyme.
  • As an example, the nucleic acid sequence of murine II-1Ra as used in the present invention is shown in the sequence listing set forth in SEQ ID NO 1. As noted above, any nucleic acid sequence resulting in a biologically active II-1Ra protein of any mammalian or human species can be used in the context of the present invention. Furthermore, also conserved nucleic acid sequences encoding for the same amino adds, polypeptide or protein fall under scope of the present invention. As illustrated in the prior art, II-1Ra genes of various species share a high homology among each other. Preferably, the helper-dependent adenoviral vector according to the invention contains a nucleic acid sequence (e.g. cDNA) of II-1Ra having at least 50 %, 60 %, 80 %, 90 % sequence homology with the nucleic acid sequence shown in SEQ ID NO 1, The invention also comprises biologically active nucleic acid sequences of II1-Ra or fragments thereof beside the full-length nucleic acid sequence of II1Ra.
  • The present invention further comprises a pharmaceutical composition, comprising a helper-dependent adenoviral vector containing a nucleic acid sequence encoding for human or mammalian interleukin-1 receptor antagonist (II-1Ra), left and right inverted terminal repeats (L ITR and R ITR), packaging signal and non-viral, non-coding stuffer nucleic acid sequences, wherein the expression of the human or mammalian interiaukin-1 receptor antagonist (II-1Ra) gene within synovial cells is regulated by an inflammallon-inducible promoter, for the treatment or prevention of osteoathritis. Preferred promoters as used in the context of the present invention are NF-κB promoter, interleukin 6 (II-6) promoter, interleukin-1 (II-1) promoter, tumor necrosis factor (TNF) promoter, cyclooxygenase 2 (COX-2) promoter, complement factor 3 (C3) promoter, serum amyloid A3 (SAA3) promoter, macrophage inflammatory protein-1α (MIP-1α) promoter, or hybrid constructs of the above.
  • It is a great benefit of the present invention that the adenoviral delivery and expression system specifically locates in the joints when administered intra-atrticularly. Most importantly, no measurable concentration of vector sequences could be deteced in the liver of mice treated with the adenoviral system of the invention. Therefore, II-1Ra concentrations are expected to be highest in the joints injected with the vector of the invention while no significant side effects are expected in any other organ.
  • The invention will be further illustrated in the examples following below.
  • Examples:
  • High levels of II-1Ra were measured in supernatants of synovial cells that were infected with a helper-dependent adenoviral vector (HDAd) of the invention. As shown below, the induction of inflammation with lipopolysaccharide (LPS) led to a dramatic increase of II-1Ra concentration as compared with uninduced samples. No II-1Ra was detected in non-infected samples (mock) or samples infected with a control vector (HDAd-GFP). The experiments demonstrate that cells infected with HDAd-mll-1Ra can produce high levels of II-1-RA. It further shows that II-1Ra is efficiently secreted from those cells, and that inflammatory conditions activate the NF-κB5-ELAM promoter leading to increased II-1Ra levels.
  • Production of the helper-dependent adenoviral vector of the invention
  • Figure 1 shows gene maps of the HDAd vectors of the invention. The full vector sequence is shown in SEQ ID NO 2 OR SEQ ID NO 3. The only difference between the two vectors is that GQ-201 carries the equine variant of II-1Ra whereas HDAd-mll-1Ra has the murine II-1Ra variant. Both vectors contain the inflammation inducible NF-κB5-ELAM promoter upstream of the II-1Ra cDNA according to SEQ ID NO 1 as well as inverted terminal repeats (ITR) and an adenoviral packaging signal. The vectors were cloned by standard digestion/ligation reactions according to the following strategy. The luciferase cDNA in pNifty-luc, a plasmid that contains the luciferase cDNA driven by a NF-κB5-ELAM promoter, was excised with Ncol and Nhel and cDNAs for equine or murine II-1Ra were ligated into this position. The NF-κB5-ELAM promoter - murine II-1Ra or NF-κB5-ELAM promoter-equine II-1Ra cassettes were excised with NotI and PacI or EcoRI and PacI. blunted and inserted into pLPBL shuttle plasmid, which had been linearized with SalI and blunted. The NF-κB5-ELAM promoter- murine II-1Ra or NF-κB5-ELAM promoter - equine II-1Ra cassettes were then excised with AscI, which flanks both sides of the multiple cloning site, and ligated into AscI linearized pΔ28 plasmid (Toietta, G., Pastore, L., Cerullo, V., Finegold, M., Beaudet, A.L., and Lee, B. (2002). Generation of helper-dependent adenoviral vectors by homologous recombination. .), which yielded the genomic plasmids pΔ28-mll-1Ra and pΔ28-eqll-1Ra. These plasmids were digested with PmeI in order to linearize the vector, liberate the inverted terminal repeats and excise bacterial resistance genes. Vectors were rescued and amplified as described before using the helper-virus AdNG163R-2 and 116 cell factories (Palmer, D., and Ng, P. (2003). Improved system for helper-dependent adenoviral vector production. Mol Ther 8, 846-852 ;Suzuki, M.. Cela, R., Clarke, C., Bertin, T.K.. Mouriño, S., and Lee, B. (2010). Large-scale production of high-quality helper-depen dent adenoviral vectors using adherent cells in cell factories. Hum Gene Ther 21, 120-126.)
  • HDAd mediates long-term marker gene expression in joints
  • In order to determine long-term gene expression for up to one year in joints, mice were infected intra-articularly with a helper-dependent adenoviral vector of the invention (HDAd) and, for comparison, a first generation adenovirus (Ad) vector expressing firefly luciferase (luc) under the control of a CMV promoter. Luc expression was followed over time using in vivo bioluminescence imaging. Strong initial luc signals were detected three days after injection with both vectors (Figure 2A). Expression decreased with both vectors thereafter and was undetectable after one month with the first generation vector Ad-luc (Figure 2B). However, HDAd-luc luciferase expression stabilized at day 10 and has been at this level for 380 days.
  • HDAd transduces synovial cells following intraarticular injection
  • To evaluate HDAd transduction in mouse joints in detail, mice were injected intra-articularly with a LacZ expressing HDAd. Strong LacZ expression was seen in the synovium, however, no expression could be observed in chondrocytes (Figure 3). The inventors also analyzed the liver of these animals to assess whether virus escapes from the joints or is spilled during the injection. Most importantly, no detectable vector concentrations over background could be measured by quantitative PCR (data not shown). Therefore, the vector specifically locates in tne joints and remains there, which is of great benefit in the treatment or prevention of an osteoarthritic condition since it suggests minimal side effects.
  • HDAd-II-1Ra infected cells secrete II-1Ra
  • An HDAd expressing II-1Ra under the control of the inflammation inducible NF-κB5-ELAM promoter was generated and its functionality was tested in vitro. High levels of II-1Ra were measured in the supernatant of HDAd-II-1Ra infected cells on day 3 (Figure 4). Induction of inflammation with lipopolysaccharide (LPS) led to a dramatic increase of II-1Ra concentration compared with uninduced samples. No II-1Ra was detected in non-infected samples (mock) or samples infected with a control vector (HDAd-GFP).
  • HDAd-II-1Ra prevents the development of OA in mice
  • To assess whether an HDAd expressing II-1Ra is able to prevent the development of OA, knee joints of mice were injected intra-articularly with HDAd-II-1Ra or a GFP expressing control vector (HDAd-GFP). Two days after injection, cruciate ligament transection was performed to induce OA development This osteoarthritis model was developed in Dr. Brendan Lee's research group and validated in several experiments (Ruan. Z., Dawson, B., Jiang M.M., Gannon, F., Heggeness, M., Lee, B. (2012). Quantitative volumetric imaging of murine osteoarthritic cartilage by phase contrast micro-computed tomography, submitted ). The model involves transection of anterior and posterior cruciate ligaments of the knee joints, which leads to development of severe OA. Mice were sacrificed one month after OA induction and joints were prepared histologically and stained with Safranin O. The development of OA was scored by a blinded pathologist according to OARSI (Osteoarthritis Research Society International) standard (assignment of scores on a scale of 1-6, 1: no signs of OA at all, 6: maximum OA). HoAd-II-1Ra treated joints had significantly lower OA scores than HDAd-GFP treated or untreated joints, suggesting that HDAd-II1Ra prevented the development of OA (Figure 5). The control vector HDAd-GFP did not seem to have any effect on the development of OA since the average OA score was comparable to the score of the untreated group.
  • HDAd-mII-Ra treats OA in a murine model of the disease
  • The efficacy of HDAd-mII-1Ra in the treatment of OA was evaluated in the murine disease model described above. The model was used to assess whether HDAd-II1-Ra can efficiently treat OA. Therefore, OA was induced by cruciate ligament transection (except in the untransected group) and OA was allowed to develop for two weeks. HDAd-II-1Ra, the control vector (HDAd-GFP) or vehicle was then injected and mice were sacrificed to analyze the joints another six weeks later. HDAd-GFP treated and uninjected mice developed OA to the same extent with an average score of approximately 4.5 (Figure 6A). However, HDAd-II-1Ra treated mice had significantly lower OA scores compared with HDAd-GFP and mock treated. No significant difference was found between HDAd-II-1Ra and untransected (OA-free) mice suggesting efficient treatment of the disease or its prevention. The inventors further evaluated the joints in this experiment by micro computer tomography (µCT) analysis. This technique combines high resolution (down to 0.5 micron) x-ray CT scanning with phase contrast optics, which enables visualization of cartilage in small animal joints. Three-dimensional reconstruction of joints and computational tissue analysis tools can be used to quantify several cartilage parameters such as volume and surface area. HDAd-II-1Ra treated joints demonstrated significantly higher cartilage volume compared with HDAd-GFP and mock treated joints (Figure 6B). No significant difference was seen between the HDAd-11-1Ra and untransected (OA-free) groups. Furthermore, cartilage surface area was significantly larger in HDAd-II-1Ra treated mice compared with HDAd-GFP and mock groups (Figure 6C), while no significant difference was seen between HDAd-II-1Ra and untransected (OA-free) joints.
  • Figure Legends: Figure 1
  • The Figure shows a basic gene map of the helper-dependent adenoviral vector of the invention. The vector backbone consists of the left and right inverted terminal repeats (ITR), adenoviral packaging signal (ψ) and non-coding, non-viral stuffer sequences (remaining unmarked sequence between ITRs). The cDNA of murine II-1Ra is cloned between the viral left and right ITRs of the used adenoviral vector. The gene of II1-Ra is controlled by inflammation-inducible NF-κB5-ELAM promoter.
  • Figure 2
  • A. Helper-dependent and first generation adenoviral vectors mediate the same level of marker gene expression. Mice were injected intra-articularly with 108 virus particles (VP) of a luciferase expressing helper-dependent (HDAd-luc) or a respective first generation (Ad-luc) adenoviral vector. Three days later mice were imaged using IVIS 200 series Imaging system (Caliper Life Sciences, Hopkintom MA). Strong bioluminescence signals were detected in the joints injected with both HDAd-luc and Ad-luc adenoviral vector. Both knee joints of four mice per group were injected; representative pictures of two mice of each group are shown.
  • B. Helper-dependent adenoviral vector mediates long-term marker gene expression in joints. Luciferase expression of the mice described in A was followed by repeated bioluminescence imaging and quantified using Living Image 2.5 software (Caliper Life Sciences). Expression decreased and was undetectable by 30 days with the first generation adenoviral vector (Ad-lue). With the helper-dependent adenoviral vector (HDAd-luc) expression also declined but plateaued at 10 days and has been around this level for 380 days.
  • Figure 3
  • Helper-dependent adenoviral vector infects synoviocytes efficiently. Mice were Injected intra-articularly with 108 VP of a LacZ expressing HDAd. One day later, mice were sacrificed and LacZ slaining on sectioned joints was performed. Strong expression (dark blue staining) was seen in the synovium while no staining could be observed in chondrocytes. Right picture is a higher magnification photograph (40x) of the framed area in the left picture (5x).
  • Figure 4
  • Cells infected with HDAd-II-1Ra produce large amounts of II-1Ra. Human embryonic kidney cells (HEK293) were infected with 100 VP/cell of HDAd-II-1Ra, HDAd-GFP or mock. Two days later II-1Ra ELISA was performed with cell culture supernatant. Concentrations of about 700 pg/ml were measured for HDAd-II-1Ra infected cells, whereas no II-1Ra was detectable in the supernatant of HDAD-GFP or mock infected cells. To induce an inflammatory reaction, lipopolysaccharides (LPS, 100 ug/ml) were added to half of the samples and II-1Ra concentrations were again determined one day later (day 4). Levels in HDAd-II-1Ra samples increased to about 1600 pg/ml whereas uninduced cells produced less II-1Ra compared to the previous day. No II-1Ra expression was detected in any of the control samples (HDAd-GFP and mock).
  • Figure 5
  • HDAd-II-1Ra prevents the development of OA. Mice were injected intra-articularly into the knee joints with 108 VP of HDAd-II-1Ra, HDAd-GFP or mock and OA was induced by cruciate ligament transduction two days later. Mice were sacrificed after 4 weeks and joints were histologically prepared, sectioned and stained with Safranin O. A blinded pathologist evaluated the level of OA according to OARSI (Osteoarthritis Research Society International) standards (assignment of scores on a scale of 1-6, 1: no signs of OA at all, 6: maximum OA). Mice treated with HDAd-II-1Ra had significantly lower OA scores compared with mice treated with HDAd-GFP and mock. (* indicates significant difference: p < 0.05 by one-way ANOVA: n =10 joints par group).
  • Figure 6
  • HDAd-II-1Ra efficiently treats OA in mice.
    A. HDAd-II-1Ra treated joints have significantly lower OA scores compared to controls. OA was induced in mouse knee joints by cruciate ligament transection and the disease was allowed to develop, Two weeks after transection, mice were injected intra-articularly with 108 VP of HDAd-II-1Ra, HDAd-GFP or mock. Mice were sacrificed 6 weeks later and joints were histologically prepared, sectioned and stained with Safranin O. A blinded pathologist evaluated the level of OA according to OARSI (Osteoarthritis Research Society International) standard (assignment of scores on a scale of 1-6, 1: no signs of OA at all, 6: maximum OA). Mice treated with HDAd-II-1Ra had significantly lower OA scores compared with mice treated with HDAd-GFP and mock. No significant difference was found between the HDAd-II-1Ra group and age matched, untransected (no OA induction) mice. (* indicates significant difference: p < 0.05 by one-way ANOVA; n = 8 joints per group). B. HDAd-II-1Ra treated joints demonstrate significantly higher cartilage volume compared to controls. Whole knee joints of mice treated the same way as described above were fixed in electron microscopy fixative and embedded in paraffin. Samples were scanned using X-radia microXCT scanner (Xradia, Pleasanton, CA, USA) and was visualized at 4 micron resolution. Computational 3D reconstruction of joints was performed and cartilage volume and surface area were quantified semi-automatically using TRI BON software (RATOC System Engineering Tokyo, Japan). Significantly higher cartilage volume was measured in HDAd-II-1Ra treated joints in comparison to controls. HDAd-II-1Ra joints had similar volumes as untransected (healthy) joints. (* indicates significant difference: p < 0.05, one-way ANOVA, n = 6 joints/group).
  • C. HDAd-II-1Ra treated joints demonstrate significantly larger cartilage surface area compared to controls. Cartilage surface area was measured as described above. HDAd-II-1Ra treatment resulted in significantly higher cartilage surface area compared to controls. Surface area of HDAd-II-1Ra treated joints was similar to that of untransected (healthy) controls. (* indicates significant difference: p < 0.05, one-way ANOVA, n = 6 joints/group).
    Figure imgb0001
    Figure imgb0002
    Figure imgb0003
    Figure imgb0004
    Figure imgb0005
    Figure imgb0006
    Figure imgb0007
    Figure imgb0008
    Figure imgb0009
    Figure imgb0010
    Figure imgb0011
    Figure imgb0012
    Figure imgb0013
    Figure imgb0014
    Figure imgb0015
    Figure imgb0016
    Figure imgb0017
    Figure imgb0018
    Figure imgb0019
    Figure imgb0020
    Figure imgb0021
    Figure imgb0022
    Figure imgb0023
    Figure imgb0024
    Figure imgb0025
    Figure imgb0026
    Figure imgb0027
    Figure imgb0028
    Figure imgb0029
    Figure imgb0030

Claims (15)

  1. An adenoviral-based biological delivery and expression system for use in the treatment or prevention of osteoathritis in human or mammalian joints by long-term inducible gene expression of human or mammalian interleukin-1 receptor antagonist (II-1Ra) in synovial cells, comprising a helper-dependent adenoviral vector containing a nucleic acid sequence encoding for human or mammalian interleukin-1 receptor antagonist (II-1Ra), left and right inverted terminal repeats (L ITR and R ITR), the adenoviral packaging signal and non-viral, non-coding stuffer nucleic acid sequences, wherein the expression of the human or mammalian interleukin-1 receptor antagonist (II-1Ra) gene within synovial cells is regulated by an inflammation-inducible promoter.
  2. The adenoviral-based biological delivery and expression system according to claim 1, wherein the inflammation-inducible promoter is selected from the group consisting of NF-κB promoter, interleukin 6 (II-6) promoter, interieukin-1 (II-1) promoter, tumor necrosis factor (TNF) promoter, cyclooxygenase 2 (COX-2) promoter, complement factor 3 (C3) promoter, serum amyloid A3 (SAA3) promoter, macrophage inflammatory protein-1α (MIP-1α) promoter, or hybrid constructs of the above.
  3. The adenoviral-based biological delivery and expression system according to claim 1 or claim 2, wherein the helper-dependent adenoviral vector comprises a nucleic acid sequence set forth in SEQ ID NO 2 or SEQ ID NO 3, or a biologically effective part thereof.
  4. The adenoviral-based biological delivery and expression system according to any one of the preceding claims, wherein the mammalian interieukin-1 receptor antagonist (II-1Ra) is selected from the group consisting of murine II-1Ra, equine II-1Ra, canine II-1Ra, cat II-1Ra, rabbit II-1Ra, hamster II-1Ra, bovine II-1Ra, camel II-1Ra or their homologs in other mammalian species.
  5. The adenoviral-based biological delivery and expression system according to any one of the preceding claims, wherein the helper-dependent adenoviral vector further comprises a marker gene that allows monitoring of the vector genome in the synovial cells.
  6. The adenoviral-based biological delivery and expression system according to any one of the preceding claims, wherein the helper-dependent vector comprises a nucleic acid sequence set forth in SEQ ID NO 1 or a conserved sequence thereof encoding for the same amino acids.
  7. The adenoviral-based biological delivery and expression system according to any one of the preceding claims, wherein the helper-dependent vector has at least 50 %, 60 %, 80%, 90 % sequence homology with the nucleic acid sequence set forth in SEQ ID NO 1.
  8. A pharmaceutical composition, comprising a helper-dependent adenoviral vector containing a nucleic acid sequence encoding for human or mammalian interieukin-1 receptor antagonist (II-1Ra), left and right inverted terminal repeats (L ITR and R ITR), an adenoviral packaging signal and non-viral, non-coding stuffer nucleic acid sequences, wherein the expression of the human or mammalian interleukin-1 receptor antagonist (II-1Ra) gene within synovial cells is regulated by an inflammation-inducible promoter, for the treatment or prevention of osteoathritis.
  9. The pharmaceutical composition according to claim 8, wherein the inflammation-inducible promoter is selected from the group consisting of NF-κB promoter, interleukin 6 (II-6) promoter, interleukin-1 (II-1) promoter, tumor necrosis factor (TNF) promoter, cyclooxygenase 2 (COX-2) promoter, complement factor 3 (C3) promoter, serum amyloid A3 (SAA3) promoter, macrophage inflammatory protein-1α (MIP-1α) promoter or hybrid constructs of the above.
  10. The pharmaceutical composition according to claim 8 or claim 9, wherein the helper-dependent adenoviral vector comprises a nucleic acid sequence set forth in SEQ ID NO 2 or SEQ ID NO 3, or a biologically effective part thereof.
  11. The pharmaceutical composition according to any one of the preceding claims, wherein the mammalian interleukin-1 receptor antagonist (II-1Ra) is selected from the group consisting of murine II-1Ra, equine II-1Ra, canine II-1Ra, cat II-1Ra, rabbit II-1Ra, hamster II-1Ra, bovine II-1Ra, camel II-1Ra or their homologs in other mammalian species.
  12. The pharmaceutical composition according to any one of the preceding claims, wherein the helper-dependent adenoviral vector further comprises a marker gene that allows monitoring of the vector genome in the synovial cells.
  13. The pharmaceutical composition according to any one of the preceding claims, wherein the helper-dependent vector comprises a nucleic acid sequence set forth in SEQ ID NO 1 or a conserved sequence thereof encoding for the same amino acids.
  14. The pharmaceutical composition according to any one of the preceding claims, wherein the helper-dependent vector has at least 50 %, 60 %, 80%, 90 % sequence homology with the nucleic acid sequence set forth in SEQ ID NO 1.
  15. Use of an adenoviral-based biological delivery and expression system according to any one of claims 1 to 7 for expressing interleukin-1 receptor antagonist (II-1Ra) in synovial cells.
EP20120000703 2012-02-02 2012-02-02 Adenoviral-based biological delivery and expression system for use in the treatment of osteoarthritis Active EP2623604B8 (en)

Priority Applications (16)

Application Number Priority Date Filing Date Title
EP20120000703 EP2623604B8 (en) 2012-02-02 2012-02-02 Adenoviral-based biological delivery and expression system for use in the treatment of osteoarthritis
DK12000703.4T DK2623604T3 (en) 2012-02-02 2012-02-02 Adenovirus release and biological expression system for use in the treatment of osteoarthritis
PL12000703T PL2623604T3 (en) 2012-02-02 2012-02-02 Adenoviral-based biological delivery and expression system for use in the treatment of osteoarthritis
ES12000703.4T ES2534782T3 (en) 2012-02-02 2012-02-02 Biological system of administration and expression based on adenovirus for use in the treatment of osteoarthritis
US14/375,351 US10301647B2 (en) 2012-02-02 2013-01-23 Adenoviral-based biological delivery and expression system for use in the treatment of osteoarthritis
EA201491457A EA033004B1 (en) 2012-02-02 2013-01-23 HELPER-DEPENDENT ADENOVIRAL VECTOR FOR DELIVERY AND LONG-TERM EXPRESSION OF INTERLEUKIN-1 RECEPTOR ANTAGONIST (Il-1Ra) AND USE THEREOF FOR THE TREATMENT AND PREVENTION OF OSTEOARTHRITIS
IN1682KON2014 IN2014KN01682A (en) 2012-02-02 2013-01-23
JP2014555328A JP6340320B2 (en) 2012-02-02 2013-01-23 Pharmaceutical composition comprising a helper-dependent adenoviral vector used in the treatment of osteoarthritis
CN201380007814.1A CN104245941B (en) 2012-02-02 2013-01-23 For treating Biodelivery based on adenovirus and the expression system of osteoarthritis
CA2861408A CA2861408C (en) 2012-02-02 2013-01-23 Adenoviral-based biological delivery and expression system for use in the treatment of osteoarthritis
PCT/IB2013/000198 WO2013114199A1 (en) 2012-02-02 2013-01-23 Adenoviral-based biological delivery and expression system for use in the treatment of osteoarthritis
AU2013213873A AU2013213873B2 (en) 2012-02-02 2013-01-23 Adenoviral-based biological delivery and expression system for use in the treatment of osteoarthritis
JP2018016342A JP2018075035A (en) 2012-02-02 2018-02-01 Adenovirus-based biological delivery and expression systems used for treatment of osteoarthritis
US16/398,691 US20190376080A1 (en) 2012-02-02 2019-04-30 Adenoviral-based biological delivery and expression system for use in the treatment of osteoarthritis
US17/497,045 US20220073948A1 (en) 2012-02-02 2021-10-08 Adenoviral-based biological delivery and expression system for use in the treatment of osteoarthritis
US17/855,322 US20220348963A1 (en) 2012-02-02 2022-06-30 Adenoviral- based biological delivery and expression system for use in the treatment of osteoarthritis

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
EP20120000703 EP2623604B8 (en) 2012-02-02 2012-02-02 Adenoviral-based biological delivery and expression system for use in the treatment of osteoarthritis

Publications (3)

Publication Number Publication Date
EP2623604A1 true EP2623604A1 (en) 2013-08-07
EP2623604B1 EP2623604B1 (en) 2015-02-25
EP2623604B8 EP2623604B8 (en) 2015-04-22

Family

ID=47891791

Family Applications (1)

Application Number Title Priority Date Filing Date
EP20120000703 Active EP2623604B8 (en) 2012-02-02 2012-02-02 Adenoviral-based biological delivery and expression system for use in the treatment of osteoarthritis

Country Status (12)

Country Link
US (4) US10301647B2 (en)
EP (1) EP2623604B8 (en)
JP (2) JP6340320B2 (en)
CN (1) CN104245941B (en)
AU (1) AU2013213873B2 (en)
CA (1) CA2861408C (en)
DK (1) DK2623604T3 (en)
EA (1) EA033004B1 (en)
ES (1) ES2534782T3 (en)
IN (1) IN2014KN01682A (en)
PL (1) PL2623604T3 (en)
WO (1) WO2013114199A1 (en)

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2948553B1 (en) * 2013-01-25 2020-04-01 Baylor College Of Medicine A helper-dependent adenoviral gene therapy delivery and expression system
GB201410314D0 (en) * 2014-06-10 2014-07-23 Advanced Risc Mach Ltd Display controller
CN107636146A (en) * 2015-03-02 2018-01-26 同生公司 It is engineered to treat the bacterium of the disease for the gastrointestinal mucosal barrier benefited from the alimentary canal inflammation of reduction and/or tightened up
WO2017123934A1 (en) 2016-01-13 2017-07-20 Merial, Inc. Recombinant aav vectors expressing osteoprotective genes, including has2 and lubricin, useful in the treatment of osteoarthritis and related joint conditions in mammals
AU2017313844B2 (en) 2016-08-19 2023-10-12 University Of Florida Research Foundation, Incorporated Methods and compositions for treating conditions using recombinant self-complementary adeno-associated virus
WO2018071295A1 (en) 2016-10-10 2018-04-19 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for inducible production of anti-inflammatory cytokines
CN110225923A (en) * 2016-12-07 2019-09-10 佛罗里达大学研究基金会有限公司 IL-1Ra cDNA
JP7260173B2 (en) 2017-04-21 2023-04-18 ベイラー カレッジ オブ メディスン Oncolytic virus therapy and immunotherapy
EP4031192A4 (en) * 2019-09-18 2023-10-25 Pacira Therapeutics, Inc. Effective dosages of an adenoviral-based biological delivery and expression system for use in the treatment of osteoarthritis in humans, and compositions comprising the same
JPWO2022234844A1 (en) * 2021-05-06 2022-11-10
KR20240052793A (en) * 2021-08-24 2024-04-23 파시라 테라퓨틱스, 인크. IL-1Ra gene therapy for intervertebral disc degeneration

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5747072A (en) 1993-07-30 1998-05-05 University Of Michigan Adenoviral-mediated gene transfer to synovial cells in vivo
US20030091536A1 (en) 1999-09-07 2003-05-15 Colorado State University Research Foundation In vivo treatment of joint disease using interleukin-1

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2806418B1 (en) * 2000-03-14 2004-07-16 Aventis Pharma Sa HYBRID INFLAMMATION INDUCIBLE PROMOTERS, VECTORS CONTAINING THEM, AND USES
WO2002083080A2 (en) 2001-04-17 2002-10-24 Genetix Pharmaceuticals, Inc. Method of treating arthritis using lentiviral vectors in gene therapy
ES2569917T3 (en) * 2005-06-21 2016-05-13 Xoma (Us) Llc Antibodies and fragments thereof that bind to IL-1beta
US8298533B2 (en) * 2008-11-07 2012-10-30 Medimmune Limited Antibodies to IL-1R1
WO2010127275A1 (en) * 2009-05-01 2010-11-04 Oregon Health & Scince University Method of expanding human hepatocytes in vivo
CA2763161A1 (en) * 2009-05-29 2010-12-02 Xoma Technology Ltd. Cardiovascular related uses of il-1.beta. antibodies and binding fragments thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5747072A (en) 1993-07-30 1998-05-05 University Of Michigan Adenoviral-mediated gene transfer to synovial cells in vivo
US20030091536A1 (en) 1999-09-07 2003-05-15 Colorado State University Research Foundation In vivo treatment of joint disease using interleukin-1

Non-Patent Citations (22)

* Cited by examiner, † Cited by third party
Title
BAKKER A C ET AL: "C3-Tat/HIV-regulated intraarticular human interleukin-1 receptor antagonist gene therapy results in efficient inhibition of collagen-induced arthritis superior to cytomegalovirus-regulated expression of the same transgene.", ARTHRITIS AND RHEUMATISM JUN 2002 LNKD- PUBMED:12115199, vol. 46, no. 6, June 2002 (2002-06-01), pages 1661 - 1670, XP002675514, ISSN: 0004-3591 *
BROOKS PM.: "Impact of osteoarthritis on individuals and society: how much disability? Social consequences and health economic implications.", CURR OPIN RHEUMATOL, vol. 14, 2002, pages 573 - 577
BRUNETTI-PIERRI, N. ET AL.: "Adenoviral Vectors", vol. 19, 1 May 2011, AMERICAN SOCIETY OF GENE & CELL THERAPY, article "Multi-Year Transgene Expression in Nonhuman Primates Following Hepatic Transduction with Helper-Dependent"
CARON JP ET AL.: "Chondroprotective effect of intraarticular injections of interleukin-1 receptor antagonist in experimental osteoarthritis. Suppression of collagenase-1 expression.", ARTHRITIS RHEUM, vol. 39, 1996, pages 1535 - 1544
CARON JP; GENOVESE, RL.: "Diagnosis and Management of Lameness in the Horse", 2003, SAUNDERS, article "Principals and practices of joint disease treatment", pages: 746 - 764
D.D. FRISBIE; S.C. GHIVIZZANI; P.D. ROBBINS; C.H. EVANS; C.W. MCLLWRAITH, GENE THER, vol. 9, 2002, pages 12 - 20
DINARELLO CA.: "The Cytokine Handbook.", 2003, ACADEMIC PRESS, article "Interleukin-1 family.", pages: 643 - 668
EVANS CH; ROBBINS PD.: "Gene Therapeutics: Methods and Applications of Direct Gene Transferm", 1994, BIRKHAUSER, article "Gene therapy for arthritis", pages: 320 - 343
EVANS, C.H.; GOUZE, J.N.; GOUZE, E.; ROBBINS, P.D.; GHIVIZZANI, S.C.: "Osteoarthritis gene therapy.", GENE THER, vol. 11, 2004, pages 379 - 389
KHOURY M ET AL: "Inflammation-inducible anti-TNF gene expression mediated by intra-articular injection of serotype 5 adeno-associated virus reduces arthritis.", THE JOURNAL OF GENE MEDICINE JUL 2007 LNKD- PUBMED:17514770, vol. 9, no. 7, July 2007 (2007-07-01), pages 596 - 604, XP002675517, ISSN: 1099-498X *
MATTHEWS, G.L.; HUNTER, D.J.: "Emerging drugs for osteoarthritis.", EXPERT OPIN. EMERGING DRUGS, 2011, pages 1 - 13
MITANI, K.; GRAHAM, F.L.; CASKEY, C.T.; KOCHANEK, S.: "Rescue, propagation, and partial purification of a helper virus-dependent adenovirus vector.", PROC NATL ACAD SCI USA, vol. 92, 1995, pages 3854 - 3858
PALMER DONNA ET AL: "Improved system for helper-dependent adenoviral vector production.", MOLECULAR THERAPY : THE JOURNAL OF THE AMERICAN SOCIETY OF GENE THERAPY NOV 2003 LNKD- PUBMED:14599819, vol. 8, no. 5, November 2003 (2003-11-01), pages 846 - 852, XP002675515, ISSN: 1525-0016 *
PALMER, D.; NG, P.: "Improved system for helper-dependent adenoviral vector production", MOL THER, vol. 8, 2003, pages 846 - 852
PALMER, D.; NG, P.: "Improved system for helper-dependent adenoviral vector production.", MOL THER, vol. 8, 2003, pages 846 - 852
PARKS, R.J. ET AL.: "A Helper-dependent adenovirus vector system: removal of helper virus by Cre-mediated excision of the viral packaging signal.", PROC NATL ACAD SCI USA, vol. 93, 1996, pages 13565 - 13570
PARKS, R.J.: "Improvements in adenoviral vector technology: overcoming barriers for gene therapy.", CLIN. GENET., vol. 58, 2000, pages 1 - 11
RUAN, Z.; DAWSON, B.; JIANG M.M.; GANNON, F.; HEGGENESS, M.; LEE, B., QUANTITATIVE VOLUMETRIC IMAGING OF MURINE OSTEOARTHRITIC CARTILAGE BY PHASE CONTRAST MICRO-COMPUTED TOMOGRAPHY, 2012
SUZUKI, M.; CELA, R.; CLARKE, C.; BERTIN, T.K.; MOURINO, S; LEE, B.: "Large-scale production of high-quality helper-dependent adenoviral vectors using adherent cells in cell factories.", HUM GENE THER, vol. 21, 2010, pages 120 - 126
TOIETTA GABRIELE ET AL: "Generation of helper-dependent adenoviral vectors by homologous recombination.", MOLECULAR THERAPY : THE JOURNAL OF THE AMERICAN SOCIETY OF GENE THERAPY FEB 2002 LNKD- PUBMED:11829528, vol. 5, no. 2, February 2002 (2002-02-01), pages 204 - 210, XP002675516, ISSN: 1525-0016 *
TOIETTA, G.; PASTORE, L.; CERULLO, V.; FINEGOID, M.; BEAUDET, A.L.; LEE, B: "Generation of helper-dependent adenoviral vectors by homologous recombination.", MOL THER, vol. 5, 2002, pages 204 - 210
TOIETTA, G.; PASTORE, L.; CERULLO, V.; FINEGOLD, M.; BEAUDET, A.L.; LEE, B.: "Generation of helper-dependent adenoviral vectors by homologous recombination.", MOL THER, vol. 5, 2002, pages 204 - 210

Also Published As

Publication number Publication date
EP2623604B1 (en) 2015-02-25
US20150031083A1 (en) 2015-01-29
US20190376080A1 (en) 2019-12-12
US20220348963A1 (en) 2022-11-03
AU2013213873B2 (en) 2018-06-21
ES2534782T3 (en) 2015-04-28
PL2623604T3 (en) 2015-07-31
CA2861408C (en) 2021-10-26
US10301647B2 (en) 2019-05-28
EA201491457A1 (en) 2014-11-28
EP2623604B8 (en) 2015-04-22
JP6340320B2 (en) 2018-06-06
DK2623604T3 (en) 2015-06-01
CN104245941B (en) 2016-12-07
CA2861408A1 (en) 2013-08-08
AU2013213873A1 (en) 2014-09-25
JP2015506695A (en) 2015-03-05
US20220073948A1 (en) 2022-03-10
WO2013114199A1 (en) 2013-08-08
JP2018075035A (en) 2018-05-17
WO2013114199A8 (en) 2014-09-25
EA033004B1 (en) 2019-08-30
CN104245941A (en) 2014-12-24
IN2014KN01682A (en) 2015-10-23

Similar Documents

Publication Publication Date Title
US20220073948A1 (en) Adenoviral-based biological delivery and expression system for use in the treatment of osteoarthritis
AU2019216631B2 (en) Enhanced adoptive cell therapy
KR101810257B1 (en) Compositions and methods for treating glioblastoma gbm
US20220282280A1 (en) Helper-dependent adenoviral gene therapy delivery and expression system
DE69914382T2 (en) Using the reading frames of the adenoviral E4 region to improve gene expression
Minter et al. Adenoviral delivery of human and viral IL-10 in murine sepsis
JP7066861B2 (en) Adeno-associated virus compositions for PAH gene transfer and how to use them
Muhammad et al. Study of the efficacy, biodistribution, and safety profile of therapeutic gutless adenovirus vectors as a prelude to a phase I clinical trial for glioblastoma
CA2545815A1 (en) Adenoviral vectored tnf-a and chemoradiation to treat cancer
KR20190067279A (en) Treatment methods using adenovirus
US11813337B2 (en) Tumor-targeting synthetic adenoviruses and uses thereof
Hofherr et al. Short-term rescue of neonatal lethality in a mouse model of propionic acidemia by gene therapy
CA2501633A1 (en) Recombinant adenoviral vectors and applications thereof
Grol The evolving landscape of gene therapy strategies for the treatment of osteoarthritis
CN117203334A (en) Epigenetic gene modulation for the treatment of neurological disorders and pain
Serabian et al. Preclinical safety evaluation of biopharmaceuticals
Deol Development of helper-dependent adenovirus for gene expression in muscle
EP1198579A1 (en) Non-human adenoviral vector for gene transfer, its use and the production thereof
Sandovici Gene therapy in kidney transplantation: towards local immune suppression
Watabe T Ad35 Vector-mediated transduction F Sakurai et al
WO2004016652A2 (en) Mammaglobin promoter

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

17P Request for examination filed

Effective date: 20131121

RAX Requested extension states of the european patent have changed

Extension state: ME

Payment date: 20131121

Extension state: BA

Payment date: 20131121

RBV Designated contracting states (corrected)

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

INTG Intention to grant announced

Effective date: 20140606

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

INTG Intention to grant announced

Effective date: 20141120

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

RAP2 Party data changed (patent owner data changed or rights of a patent transferred)

Owner name: BAYLOR COLLEGE OF MEDICINE

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602012005337

Country of ref document: DE

Effective date: 20150409

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 712062

Country of ref document: AT

Kind code of ref document: T

Effective date: 20150415

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2534782

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20150428

REG Reference to a national code

Ref country code: CH

Ref legal event code: NV

Representative=s name: PATENTANWAELTE SCHAAD, BALASS, MENZL AND PARTN, CH

REG Reference to a national code

Ref country code: NL

Ref legal event code: T3

REG Reference to a national code

Ref country code: DK

Ref legal event code: T3

Effective date: 20150526

REG Reference to a national code

Ref country code: SE

Ref legal event code: TRGR

REG Reference to a national code

Ref country code: NO

Ref legal event code: T2

Effective date: 20150225

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

REG Reference to a national code

Ref country code: PL

Ref legal event code: T3

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: RS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150625

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150526

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602012005337

Country of ref document: DE

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20151126

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 5

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

REG Reference to a national code

Ref country code: AT

Ref legal event code: UEP

Ref document number: 712062

Country of ref document: AT

Kind code of ref document: T

Effective date: 20150225

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 6

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 7

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT; INVALID AB INITIO

Effective date: 20120202

Ref country code: SM

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

Ref country code: MT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160229

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: AL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: NO

Payment date: 20230208

Year of fee payment: 12

Ref country code: DK

Payment date: 20230213

Year of fee payment: 12

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: TR

Payment date: 20230202

Year of fee payment: 12

Ref country code: IT

Payment date: 20230110

Year of fee payment: 12

Ref country code: BE

Payment date: 20230117

Year of fee payment: 12

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230531

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20231214

Year of fee payment: 13

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: SE

Payment date: 20231213

Year of fee payment: 13

Ref country code: NL

Payment date: 20231215

Year of fee payment: 13

Ref country code: IE

Payment date: 20231211

Year of fee payment: 13

Ref country code: FR

Payment date: 20231212

Year of fee payment: 13

Ref country code: FI

Payment date: 20231218

Year of fee payment: 13

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: PL

Payment date: 20231212

Year of fee payment: 13

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: LU

Payment date: 20240129

Year of fee payment: 13

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: ES

Payment date: 20240306

Year of fee payment: 13

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: AT

Payment date: 20240125

Year of fee payment: 13

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: DE

Payment date: 20231205

Year of fee payment: 13

Ref country code: CH

Payment date: 20240301

Year of fee payment: 13